553
Views
37
CrossRef citations to date
0
Altmetric
Review

Wound dressings as growth factor delivery platforms for chronic wound healing

, ORCID Icon &
Pages 737-759 | Received 25 Sep 2020, Accepted 17 Dec 2020, Published online: 03 Jan 2021

References

  • Zhao A, Qin H, Fu X. What determines the regenerative capacity in animals? BioSci. 2016;66(9):735–746.
  • Iismaa SE, Kaidonis X, Nicks AM, et al. Comparative regenerative mechanisms across different mammalian tissues. Npj Regener Med. 2018;3:6.
  • Lorenz HP, Longaker MT, Perkocha LA, et al. Scarless wound repair: a human fetal skin model. Dev. 1992;114(1):253–259. Jan.
  • Gurtner GC, Werner S, Barrandon Y, et al. Wound repair and regeneration. Nat. 2008 May 15;453(7193):314–321.
  • Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Med. 2014 Dec 3;6:(265)265sr6.
  • Pugliese E, Coentro JQ, Raghunath M, et al. Wound healing and scar wars. Adv Drug Deliv Rev. 2018 Apr;129:1–3.
  • Wynn TA. Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat Rev Immunol. 2004;4(8):583–594. Aug.
  • Velnar T, Bailey T, Smrkolj V. The wound healing process: an overview of the cellular and molecular mechanisms. J Int Med Res. 2009;37(5):1528–1542. Sep-Oct.
  • Brem H, Tomic-Canic M. Cellular and molecular basis of wound healing in diabetes. J Clin Invest. 2007;117(5):1219–1222. May.
  • Xue M, Zhao R, Lin H, et al. Delivery systems of current biologicals for the treatment of chronic cutaneous wounds and severe burns. Adv Drug Deliv Rev. Apr 2018;129:219–241. .
  • Catanzano O, Boateng J. Local delivery of growth factors using wound dressings. In: Boateng J, editor. Therapeutic dressings and wound healing applications. USA: Wiley. 2020. p. 291–314. 2020/03/02.
  • Dubay DA, Franz MG. Acute wound healing: the biology of acute wound failure. Surg Clin North Am. 2003;83(3):463–481. Jun. .
  • Lazarus GS, Cooper DM, Knighton DR, et al. Definitions and guidelines for assessment of wounds and evaluation of healing. Wound Repair Regen. 1994;2(3):165–170. Jul.
  • Armstrong DG, Boulton AJM, Bus SA. Diabetic foot ulcers and their recurrence. N Engl J Med. 2017;376(24):2367–2375. 2017/06/15.
  • Zubair M, Ahmad J. Role of growth factors and cytokines in diabetic foot ulcer healing: A detailed review. Rev Endocr Metab Disord. 2019;20(2):207–217. 2019/06/01.
  • Lindley LE, Stojadinovic O, Pastar I, et al. Biology and biomarkers for wound healing. Plast Reconstr Surg. 2016;138(3 Suppl):18S–28S. Sep.
  • Trengove NJ, Stacey MC, MacAuley S, et al. Analysis of the acute and chronic wound environments: the role of proteases and their inhibitors. Wound Repair Regen. 1999;7(6):442–452. Nov-Dec.
  • McCarty SM, Percival SL. Proteases and delayed wound healing. Adv Wound Care (New Rochelle). 2013;2(8):438–447. Oct.
  • Xue M, Le NT, Jackson CJ. Targeting matrix metalloproteases to improve cutaneous wound healing. Expert Opin Ther Targets. 2006;10(1):143–155. Feb.
  • Wall IB, Moseley R, Baird DM, et al. Fibroblast dysfunction is a key factor in the non-healing of chronic venous leg ulcers. J Invest Dermatol. 2008;128(10):2526–2540. Oct.
  • Cook H, Davies KJ, Harding KG, et al. Defective extracellular matrix reorganization by chronic wound fibroblasts is associated with alterations in TIMP-1, TIMP-2, and MMP-2 activity. J Invest Dermatol. 2000;115(2):225–233. Aug.
  • Rodriguez-Menocal L, Salgado M, Ford D, et al. Stimulation of skin and wound fibroblast migration by mesenchymal stem cells derived from normal donors and chronic wound patients. Stem Cells Transl Med. 2012;1(3):221–229. Mar.
  • Stojadinovic O, Brem H, Vouthounis C, et al. Molecular pathogenesis of chronic wounds: the role of beta-catenin and c-myc in the inhibition of epithelialization and wound healing. Am J Pathol. 2005;167(1):59–69. Jul.
  • Kim H, Kim Y, Park J, et al. Recent advances in engineered stem cell-derived cell sheets for tissue regeneration. Polymers (Basel). 2019 Jan 26;11(2):209.
  • Ulrich D, Ulrich F, Unglaub F, et al. Matrix metalloproteinases and tissue inhibitors of metalloproteinases in patients with different types of scars and keloids. J Plast Reconstr Aesthet Surg. 2010;63(6):1015–1021.
  • Xue M, Jackson CJ. Extracellular matrix reorganization during wound healing and its impact on abnormal scarring. Adv Wound Care (New Rochelle). 2015 Mar 1;4(3):119–136.
  • Cubison TC, Pape SA, Parkhouse N. Evidence for the link between healing time and the development of hypertrophic scars (HTS) in paediatric burns due to scald injury. Burns. 2006;32(8):992–999. Dec.
  • Finnerty CC, Jeschke MG, Branski LK, et al. Hypertrophic scarring: the greatest unmet challenge after burn injury. Lancet. 2016 Oct 1;388(10052):1427–1436.
  • Zhu Z, Ding J, Tredget EE. The molecular basis of hypertrophic scars. Burns Trauma. 2016;4:2.
  • Lian N, Li T. Growth factor pathways in hypertrophic scars: molecular pathogenesis and therapeutic implications. Biomed Pharmacother. 2016;84:42–50.
  • Vågesjö E, Öhnstedt E, Mortier A, et al. Accelerated wound healing in mice by on-site production and delivery of CXCL12 by transformed lactic acid bacteria. Proc Nat Acad Sci (PNAS). 2018;115(8):1895–1900.
  • Tripathi S, Soni K, Agrawal P, et al. Hypertrophic scars and keloids: a review and current treatment modalities. Biomed Dermatol. 2020;4(1):11.
  • Huang C, Murphy GF, Akaishi S, et al. Keloids and hypertrophic scars: update and future directions. Plast Reconstr Surg Glob Open. 2013;1(4):e25.
  • Smith CJ, Smith JC, Finn MC. The possible role of mast cells (allergy) in the production of keloid and hypertrophic scarring. J Burn Care Rehabil. 1987;8(2):126–131.
  • Werner S, Grose R. Regulation of wound healing by growth factors and cytokines. Physiol Rev. 2003;83(3):835–870.
  • Kim HS, Sun X, Lee JH, et al. Advanced drug delivery systems and artificial skin grafts for skin wound healing. Adv Drug Deliv Rev. 2018 Dec 31.
  • Rennert RC, Rodrigues M, Wong VW, et al. Biological therapies for the treatment of cutaneous wounds: phase III and launched therapies. Expert Opin Biol Ther. 2013;13(11):1523–1541.
  • Leader B, Baca QJ, Golan DE. Protein therapeutics: a summary and pharmacological classification. Nat Rev Drug Discov. 2008;7(1):21–39.
  • Park JW, Hwang SR, Yoon IS. Advanced growth factor delivery systems in wound management and skin regeneration. Mol. 2017 Jul 27;22(8):1259.
  • Barrientos S, Brem H, Stojadinovic O, et al. Clinical application of growth factors and cytokines in wound healing. Wound Repair Regen. 2014;22(5):569–578. Sep-Oct.
  • Barrientos S, Stojadinovic O, Golinko MS, et al. Growth factors and cytokines in wound healing. Wound Repair Regen. 2008;16(5):585–601. Sep-Oct.
  • Dinh T, Braunagel S, Rosenblum BI. Growth factors in wound healing: the present and the future? Clin Podiatr Med Surg. 2015;32(1):109–119. Jan.
  • Falanga V. Chronic wounds: pathophysiologic and experimental considerations. J Invest Dermatol. 1993;100(5):721–725. 1993/05/01/.
  • Pierce GF, Tarpley JE, Tseng J, et al. Detection of platelet-derived growth factor (PDGF)-AA in actively healing human wounds treated with recombinant PDGF-BB and absence of PDGF in chronic nonhealing wounds. J Clin Invest. 1995;96(3):1336–1350. Sep.
  • Schultz GS, Wysocki A. Interactions between extracellular matrix and growth factors in wound healing. Wound Repair Regen. 2009;17(2):153–162. Mar-Apr.
  • Quatresooz P, Henry F, Paquet P, et al. Deciphering the impaired cytokine cascades in chronic leg ulcers (review). Int J Mol Med. 2003;11(4):411–418. Apr.
  • Mast BA, Schultz GS. Interactions of cytokines, growth factors, and proteases in acute and chronic wounds. Wound Repair Regen. 1996;4(4):411–420. Oct.
  • Cooper DM, Yu EZ, Hennessey P, et al. Determination of endogenous cytokines in chronic wounds. Ann Surg. Jun 1994;219(6):688–691. discussion 91–2.
  • Cianfarani F, Tommasi R, Failla CM, et al. Granulocyte/macrophage colony-stimulating factor treatment of human chronic ulcers promotes angiogenesis associated with de novo vascular endothelial growth factor transcription in the ulcer bed. Br J Dermatol. 2006;154(1):34–41. Jan.
  • Berlanga-Acosta J, Fernandez-Montequin J, Valdes-Perez C, et al. Diabetic foot ulcers and epidermal growth factor: revisiting the local delivery route for a successful outcome. Biomed Res Int. 2017;2017:2923759.
  • Gainza G, Villullas S, Pedraz JL, et al. Advances in drug delivery systems (DDSs) to release growth factors for wound healing and skin regeneration. Nanomed. 2015;11(6):1551–1573. Aug.
  • Brown GL, Nanney LB, Griffen J, et al. Enhancement of wound healing by topical treatment with epidermal growth factor. N Engl J Med. 1989 Jul 13;321(2):76–79.
  • Fang RC, Galiano RD. A review of becaplermin gel in the treatment of diabetic neuropathic foot ulcers. Biologics. 2008;2(1):1–12. Mar.
  • Steed DL. Clinical evaluation of recombinant human platelet-derived growth factor for the treatment of lower extremity diabetic ulcers. diabetic ulcer study group. J Vasc Surg. Jan 1995;21(1):71–78. discussion 79–81.
  • Wieman TJ, Smiell JM, Su Y. Efficacy and safety of a topical gel formulation of recombinant human platelet-derived growth factor-BB (becaplermin) in patients with chronic neuropathic diabetic ulcers. A phase III randomized placebo-controlled double-blind study. Diabetes Care. 1998;21(5):822–827. May.
  • Margolis DJ, Bartus C, Hoffstad O, et al. Effectiveness of recombinant human platelet-derived growth factor for the treatment of diabetic neuropathic foot ulcers. Wound Repair Regen. 2005;13(6):531–536. Nov-Dec.
  • Fernandez-Montequin JI, Betancourt BY, Leyva-Gonzalez G, et al. Intralesional administration of epidermal growth factor-based formulation (Heberprot-P) in chronic diabetic foot ulcer: treatment up to complete wound closure. Int Wound J. 2009;6(1):67–72. Feb.
  • Akita S, Akino K, Imaizumi T, et al. Basic fibroblast growth factor accelerates and improves second-degree burn wound healing. Wound Repair Regen. 2008;16(5):635–641. Sep-Oct.
  • Yao CC, Yao P, Wu H, et al. Acceleration of wound healing in traumatic ulcers by absorbable collagen sponge containing recombinant basic fibroblast growth factor. Biomed Mater. 2006;1(1):33–37. Mar.
  • Tuyet HL, Nguyen Quynh TT, Vo Hoang Minh H, et al. The efficacy and safety of epidermal growth factor in treatment of diabetic foot ulcers: the preliminary results. Int Wound J. 2009;6(2):159–166. Apr.
  • Kozynets HP, Osadcha OI, Boiars’ka HM, et al. [Determination of clinical efficacy of REGEN-D 150 preparation for local treatment of burns]. Klin Khir. 2011;7:65–68.
  • Smiell JM. Clinical safety of becaplermin (rhPDGF-BB) gel. becaplermin studies group. Am J Surg. 1998;176(2A Suppl):68S–73S. Aug.
  • Boateng JS, Matthews KH, Stevens HN, et al. Wound healing dressings and drug delivery systems: a review. J Pharm Sci. 2008;97(8):2892–2923. Aug.
  • Boateng J, Catanzano O. advanced therapeutic dressings for effective wound healing–a review. J Pharm Sci. 2015;104(11):3653–3680. Nov.
  • Biondi M, Ungaro F, Quaglia F, et al. Controlled drug delivery in tissue engineering. Adv Drug Deliv Rev. 2008 Jan 14;60(2):229–242.
  • Fenton OS, Olafson KN, Pillai PS, et al. Advances in biomaterials for drug delivery. Adv Mater. 2018 May 7;30(29):1705328.
  • Quaglia F. Bioinspired tissue engineering: the great promise of protein delivery technologies. Int J Pharm. 2008 Dec 8;364(2):281–297.
  • Choi SM, Lee KM, Kim HJ, et al. Effects of structurally stabilized EGF and bFGF on wound healing in type I and type II diabetic mice. Acta Biomater. 2018 Jan 15;66:325–334.
  • Lao G, Yan L, Yang C, et al. Controlled release of epidermal growth factor from hydrogels accelerates wound healing in diabetic rats. J Am Podiatr Med Assoc. 2012;102(2):89–98. Mar-Apr.
  • Lin YJ, Lee GH, Chou CW, et al. Stimulation of wound healing by PU/hydrogel composites containing fibroblast growth factor-2. J Mater Chem B. 2015;3(9):1931–1941.
  • Niiyama H, Kuroyanagi Y. Development of novel wound dressing composed of hyaluronic acid and collagen sponge containing epidermal growth factor and vitamin C derivative. J Artif Organs. 2014;17(1):81–87. Mar.
  • Schneider A, Wang XY, Kaplan DL, et al. Biofunctionalized electrospun silk mats as a topical bioactive dressing for accelerated wound healing. Acta Biomater. 2009;5(7):2570–2578. Sep.
  • Lord MS, Ellis AL, Farrugia BL, et al. Perlecan and vascular endothelial growth factor-encoding DNA-loaded chitosan scaffolds promote angiogenesis and wound healing. J Control Release. 2017 Mar 28;250:48–61.
  • Gilmartin DJ, Soon A, Thrasivoulou C, et al. Sustained release of cx43 antisense oligodeoxynucleotides from coated collagen scaffolds promotes wound healing. Adv Healthc Mater. 2016;5(14):1786–1799.
  • Subbiah R, Guldberg RE. Materials science and design principles of growth factor delivery systems in tissue engineering and regenerative medicine. Adv Healthc Mater. 2019;8(1):1801000.
  • Barroso A, Mestre H, Ascenso A, et al. Nanomaterials in wound healing: from material sciences to wound healing applications. Nano Select. 2020;1(5):443–460.
  • Elsabahy M, Wooley KL. Design of polymeric nanoparticles for biomedical delivery applications. Chem Soc Rev. 2012 Apr 7;41(7):2545–2561.
  • Beckert S, Farrahi F, Aslam RS, et al. Lactate stimulates endothelial cell migration. Wound Repair Regen. 2006;14(3):321–324.
  • Trabold O, Wagner S, Wicke C, et al. Lactate and oxygen constitute a fundamental regulatory mechanism in wound healing. Wound Repair Regen. 2003;11(6):504–509.
  • Borselli C, Ungaro F, Oliviero O, et al. Bioactivation of collagen matrices through sustained VEGF release from PLGA microspheres. J Biomed Mater Res A. 2010;92A(1):94–102.
  • Matica MA, Aachmann FL, Tondervik A, et al. Chitosan as a wound dressing starting material: antimicrobial properties and mode of action. Int J Mol Sci. 2019 Nov 24;20:23.
  • Desmet CM, Preat V, Gallez B. Nanomedicines and gene therapy for the delivery of growth factors to improve perfusion and oxygenation in wound healing. Adv Drug Deliv Rev. 2018;129:262–284.
  • Kalashnikova I, Das S, Seal S. Nanomaterials for wound healing: scope and advancement. Nanomed (Lond). 2015;10(16):2593–2612.
  • Vijayan A, James PP, Nanditha CK, et al. Multiple cargo deliveries of growth factors and antimicrobial peptide using biodegradable nanopolymer as a potential wound healing system. Int J Nanomed. 2019;14:2253–2263.
  • Yang DH, Seo DI, Lee D-W, et al. Preparation and evaluation of visible-light cured glycol chitosan hydrogel dressing containing dual growth factors for accelerated wound healing. J Ind Eng Chem. 2017;53::360–370.
  • Shimizu N, Ishida D, Yamamoto A, et al. Development of a functional wound dressing composed of hyaluronic acid spongy sheet containing bioactive components: evaluation of wound healing potential in animal tests. J Biomater Sci Polym Ed. 2014;25(12):1278–1291.
  • Choi JS, Yoo HS. Pluronic/chitosan hydrogels containing epidermal growth factor with wound-adhesive and photo-crosslinkable properties. J Biomed Mater Res A. 2010;95(2):564–573.
  • Beckert S, Haack S, Hierlemann H, et al. Stimulation of steroid-suppressed cutaneous healing by repeated topical application of IGF-I: different mechanisms of action based upon the mode of IGF-I delivery. J Surg Res. 2007 May 15;139(2):217–221.
  • Picheth GF, Sierakowski MR, Woehl MA, et al. Lysozyme-triggered epidermal growth factor release from bacterial cellulose membranes controlled by smart nanostructured films. J Pharm Sci. 2014;103(12):3958–3965.
  • Xuan X, Zhou Y, Chen A, et al. Silver crosslinked injectable bFGF-eluting supramolecular hydrogels speed up infected wound healing. J Mater Chem B. 2020 Feb 21;8(7):1359–1370.
  • Hong JP, Kim YW, Lee SK, et al. The effect of continuous release of recombinant human epidermal growth factor (rh-EGF) in chitosan film on full thickness excisional porcine wounds. Ann Plast Surg. 2008;61(4):457–462.
  • Hajimiri M, Shahverdi S, Esfandiari MA, et al. Preparation of hydrogel embedded polymer-growth factor conjugated nanoparticles as a diabetic wound dressing. Drug Dev Ind Pharm. 2016 May 3;42(5):707–719.
  • Piran M, Vakilian S, Piran M, et al. In vitro fibroblast migration by sustained release of PDGF-BB loaded in chitosan nanoparticles incorporated in electrospun nanofibers for wound dressing applications. Artif Cells Nanomed Biotechnol. 2018;46(sup1):511–520.
  • Nejaddehbashi F, Hashemitabar M, Bayati V, et al. Application of polycaprolactone, chitosan, and collagen composite as a nanofibrous mat loaded with silver sulfadiazine and growth factors for wound dressing. Artif Organs. 2019;43(4):413–423.
  • Vijayan A, S A, Kumar GSV. PEG grafted chitosan scaffold for dual growth factor delivery for enhanced wound healing. Sci Rep. 2019 Dec 16;9(1):19165.
  • Sokolsky-Papkov M, Agashi K, Olaye A, et al. Polymer carriers for drug delivery in tissue engineering. Adv Drug Delliv Rev. 2007;59(4):187–206.
  • Tanaka A, Nagate T, Matsuda H. Acceleration of wound healing by gelatin film dressings with epidermal growth factor. J Vet Med Sci. 2005;67(9):909–913.
  • Ulubayram K, Nur Cakar A, Korkusuz P, et al. EGF containing gelatin-based wound dressings. Biomaterials. 2001;22(11):1345–1356.
  • Sakamoto M, Morimoto N, Ogino S, et al. Efficacy of gelatin gel sheets in sustaining the release of basic fibroblast growth factor for murine skin defects. J Surg Res. 2016;201(2):378–387.
  • Hu Y, Zhang Z, Li Y, et al. Dual-crosslinked amorphous polysaccharide hydrogels based on chitosan/alginate for wound healing applications. Macromol Rapid Commun. 2018 May 31:e1800069.
  • Shi M, Zhang H, Song T, et al. Sustainable dual release of antibiotic and growth factor from ph-responsive uniform alginate composite microparticles to enhance wound healing. ACS Appl Mater Interfaces. 2019 Jun 26;11(25):22730–22744.
  • Ribeiro MP, Morgado PI, Miguel SP, et al. Dextran-based hydrogel containing chitosan microparticles loaded with growth factors to be used in wound healing. Mater Sci Eng C Mater Biol Appl. 2013 Jul 1;33(5):2958–2966.
  • Pyun DG, Choi HJ, Yoon HS, et al. Polyurethane foam containing rhEGF as a dressing material for healing diabetic wounds: synthesis, characterization, in vitro and in vivo studies. Colloids Surf B Biointerfaces. 2015 Nov 1;135:699–706.
  • Mohandas A, Anisha BS, Chennazhi KP, et al. Chitosan-hyaluronic acid/VEGF loaded fibrin nanoparticles composite sponges for enhancing angiogenesis in wounds. Colloids Surf B Biointerfaces. 2015 Mar 1;127:105–113.
  • Kim H, Kong WH, Seong KY, et al. Hyaluronate-epidermal growth factor conjugate for skin wound healing and regeneration. Biomacromol. 2016 Nov 14;17(11):3694–3705.
  • Wang W, Lin S, Xiao Y, et al. Acceleration of diabetic wound healing with chitosan-crosslinked collagen sponge containing recombinant human acidic fibroblast growth factor in healing-impaired STZ diabetic rats. Life Sci. 2008 Jan 16;82(3–4):190–204.
  • Wang Y, Fu C, Wu Z, et al. A chitin film containing basic fibroblast growth factor with a chitin-binding domain as wound dressings. Carbohydr Polym. 2017 Oct 15;174:723–730.
  • Silva AKA, Richard C, Bessodes M, et al. Growth factor delivery approaches in hydrogels. Biomacromol. 2009;10(1):9–18.
  • He S, Shi D, Han Z, et al. Heparinized silk fibroin hydrogels loading FGF1 promote the wound healing in rats with full-thickness skin excision. Biomed Eng Online. 2019 Oct 2;18(1):97.
  • Etxabide A, Vairo C, Santos-Vizcaino E, et al. Ultra thin hydro-films based on lactose-crosslinked fish gelatin for wound healing applications. Int J Pharm. 2017 Sep 15;530(1–2):455–467.
  • Oh JS, Lee EJ. Engineered dressing of hybrid chitosan-silica for effective delivery of keratin growth factor and acceleration of wound healing. Mater Sci Eng C Mater Biol Appl. 2019;103:109815.
  • Niu Y, Li Q, Ding Y, et al. Engineered delivery strategies for enhanced control of growth factor activities in wound healing. Adv Drug Deliv Rev. 2019;146:190–208.
  • Xie Z, Paras CB, Weng H, et al. Dual growth factor releasing multi-functional nanofibers for wound healing. Acta Biomater. 2013;9(12):9351–9359.
  • Wu J, Ye J, Zhu J, et al. Heparin-based coacervate of FGF2 improves dermal regeneration by asserting a synergistic role with cell proliferation and endogenous facilitated VEGF for cutaneous wound healing. Biomacromol. 2016 Jun 13;17(6):2168–2177.
  • Liao AH, Hung CR, Chen HK, et al. Ultrasound-mediated EGF-coated-microbubble cavitation in dressings for wound-healing applications. Sci Rep. 2018 May 29;8(1):8327.
  • Li X, Ye X, Qi J, et al. EGF and curcumin co-encapsulated nanoparticle/hydrogel system as potent skin regeneration agent. Int J Nanomed. 2016;11:3993–4009.
  • Lin X, Guan X, Wu Y, et al. An alginate/poly(N-isopropylacrylamide)-based composite hydrogel dressing with stepwise delivery of drug and growth factor for wound repair. Mater Sci Eng C. 2020/10/01/ 2020;115: 111123.
  • Jazwa A, Kucharzewska P, Leja J, et al. Combined vascular endothelial growth factor-A and fibroblast growth factor 4 gene transfer improves wound healing in diabetic mice. Genet Vaccines Ther. 2010;8:6.
  • Yancopoulos GD, Davis S, Gale NW, et al. Vascular-specific growth factors and blood vessel formation. Nat. 2000 Sep 14;407(6801):242–248.
  • Choi DH, Subbiah R, Kim IH, et al. Dual growth factor delivery using biocompatible core-shell microcapsules for angiogenesis. Small. 2013;9(20):3468–3476.
  • Losi P, Briganti E, Errico C, et al. Fibrin-based scaffold incorporating VEGF- and bFGF-loaded nanoparticles stimulates wound healing in diabetic mice. Acta Biomater. 2013;9(8):7814–7821.
  • Gizaw M, Thompson J, Faglie A, et al. Electrospun fibers as a dressing material for drug and biological agent delivery in wound healing applications. Bioeng (Basel). 2018 Jan 27;5:1.
  • Zamani M, Prabhakaran MP, Ramakrishna S. Advances in drug delivery via electrospun and electrosprayed nanomaterials. Int J Nanomedicine. 2013;8:2997–3017.
  • Miguel SP, Sequeira RS, Moreira AF, et al. An overview of electrospun membranes loaded with bioactive molecules for improving the wound healing process. Eur J Pharm Biopharm. Jun 2019;139:1–22.
  • Meinel AJ, Germershaus O, Luhmann T, et al. Electrospun matrices for localized drug delivery: current technologies and selected biomedical applications. Eur J Pharm Biopharm. 2012;81(1):1–13.
  • Son YJ, Kim WJ, Yoo HS. Therapeutic applications of electrospun nanofibers for drug delivery systems. Arch Pharm Res. 2014;37(1):69–78.
  • Chen S, Liu B, Carlson MA, et al. Recent advances in electrospun nanofibers for wound healing. Nanomed (Lond). 2017;12(11):1335–1352.
  • Liu M, Duan XP, Li YM, et al. Electrospun nanofibers for wound healing. Mater Sci Eng C Mater Biol Appl. 2017 Jul 1;76:1413–1423. doi: 10.1016/j.msec.2017.03.034.
  • Sebe I, Szabo P, Kallai-Szabo B, et al. Incorporating small molecules or biologics into nanofibers for optimized drug release: A review. Int J Pharm. 2015 Oct 15;494(1):516–530.
  • Choi JS, Leong KW, Yoo HS. In vivo wound healing of diabetic ulcers using electrospun nanofibers immobilized with human epidermal growth factor (EGF). Biomaterials. 2008;29(5):587–596.
  • Dwivedi C, Pandey I, Pandey H, et al. In vivo diabetic wound healing with nanofibrous scaffolds modified with gentamicin and recombinant human epidermal growth factor. J Biomed Mater Res A. 2018;106(3):641–651.
  • Jiang H, Wang L, Zhu K. Coaxial electrospinning for encapsulation and controlled release of fragile water-soluble bioactive agents. J Control Release. 2014 Nov 10;193:296–303.
  • Ji W, Yang F, van den Beucken JJ, et al. Fibrous scaffolds loaded with protein prepared by blend or coaxial electrospinning. Acta Biomater. 2010;6(11):4199–4207.
  • Choi JS, Choi SH, Yoo HS. Coaxial electrospun nanofibers for treatment of diabetic ulcers with binary release of multiple growth factors. J Mater Chem. 2011;21(14):5258–5267.
  • Lai HJ, Kuan CH, Wu HC, et al. Tailored design of electrospun composite nanofibers with staged release of multiple angiogenic growth factors for chronic wound healing. Acta Biomater. 2014;10(10):4156–4166.
  • McClellan P, Landis WJ. Recent applications of coaxial and emulsion electrospinning methods in the field of tissue engineering. Biores Open Access. 2016;5(1):212–227.
  • Zhang C, Feng F, Zhang H. Emulsion electrospinning: fundamentals, food applications and prospects. Trends Food Sci Tech. 2018;80:175–186.
  • Yang Y, Xia T, Zhi W, et al. Promotion of skin regeneration in diabetic rats by electrospun core-sheath fibers loaded with basic fibroblast growth factor. Biomaterials. 2011;32(18):4243–4254.
  • Garcia-Orue I, Gainza G, Gutierrez FB, et al. Novel nanofibrous dressings containing rhEGF and Aloe vera for wound healing applications. Int J Pharm. 2017 May 25;523(2):556–566.
  • Norouzi M, Shabani I, Ahvaz HH, et al. PLGA/gelatin hybrid nanofibrous scaffolds encapsulating EGF for skin regeneration. J Biomed Mater Res A. 2014 Oct 24;103(7);2225-2235.
  • Wang Z, Qian Y, Li L, et al. Evaluation of emulsion electrospun polycaprolactone/hyaluronan/epidermal growth factor nanofibrous scaffolds for wound healing. J Biomater Appl. 2016;30(6):686–698.
  • Zhao Q, Lu WW, Wang M. Modulating the release of vascular endothelial growth factor by negative-voltage emulsion electrospinning for improved vascular regeneration. Mater Lett. 2017;193:1–4.
  • Mouthuy PA, Groszkowski L, Ye H. Performances of a portable electrospinning apparatus. Biotechnol Lett. 2015;37(5):1107–1116.
  • Xu SC, Qin CC, Yu M, et al. A battery-operated portable handheld electrospinning apparatus. Nanoscale. 2015 Aug 7;7(29):12351–12355.
  • Yan X, Yu M, Zhang LH, et al. A portable electrospinning apparatus based on a small solar cell and a hand generator: design, performance and application. Nanoscale. 2016 Jan 7;8(1):209–213.
  • Brown TD, Dalton PD, Hutmacher DW. Direct writing by way of melt electrospinning. Adv Mater. 2011 Dec 15;23(47):5651–5657.
  • Ristovski N, Bock N, Liao S, et al. Improved fabrication of melt electrospun tissue engineering scaffolds using direct writing and advanced electric field control. Biointerphases. 2015 Mar 25;10(1):011006.
  • Kim G, Son J, Su A P, et al. Hybrid process for fabricating 3d hierarchical scaffolds combining rapid prototyping and electrospinning. Macromol Rapid Commun. 2008;29(19):1577–1581.
  • Ross R, Glomset J, Kariya B, et al. A platelet-dependent serum factor that stimulates the proliferation of arterial smooth muscle cells in vitro. Proc Natl Acad Sci U S A. 1974;71(4):1207–1210.
  • Mazzucco L, Borzini P, Gope R. Platelet-derived factors involved in tissue repair-from signal to function. Transfus Med Rev. 2010;24(3):218–234.
  • Bhanot S, Alex JC. Current applications of platelet gels in facial plastic surgery. Facial Plast Surg. 2002;18(1):27–33.
  • Foster TE, Puskas BL, Mandelbaum BR, et al. Platelet-rich plasma: from basic science to clinical applications. Am J Sports Med. 2009;37(11):2259–2272.
  • Saucedo JM, Yaffe MA, Berschback JC, et al. Platelet-rich plasma. J Hand Surg Am. Mar 2012;37(3):587–589.
  • Chicharro-Alcantara D, Rubio-Zaragoza M, Damia-Gimenez E, et al. Platelet rich plasma: new insights for cutaneous wound healing management. J Funct Biomater. 2018 Jan 18;9:1.
  • Eppley BL, Woodell JE, Higgins J. Platelet quantification and growth factor analysis from platelet-rich plasma: implications for wound healing. Plast Reconstr Surg. 2004;114(6):1502–1508.
  • Roukis TS, Zgonis T, Tiernan B. Autologous platelet-rich plasma for wound and osseous healing: a review of the literature and commercially available products. Adv Ther. 2006;23(2):218–237.
  • Bir SC, Esaki J, Marui A, et al. Angiogenic properties of sustained release platelet-rich plasma: characterization in-vitro and in the ischemic hind limb of the mouse. J Vasc Surg. 2009;50(4):870–79 e2.
  • Mendes BB, Gomez-Florit M, Babo PS, et al. Blood derivatives awaken in regenerative medicine strategies to modulate wound healing. Adv Drug Deliv Rev. 2018;129:376–393.
  • Marx RE. Platelet-rich plasma: evidence to support its use. J Oral Maxillofac Surg. 2004;62(4):489–496.
  • Naik B, Karunakar P, Jayadev M, et al. Role of platelet rich fibrin in wound healing: A critical review. J Conserv Dent. 2013;16(4):284–293.
  • Dougherty EJ. An evidence-based model comparing the cost-effectiveness of platelet-rich plasma gel to alternative therapies for patients with nonhealing diabetic foot ulcers. Adv Skin Wound Care. 2008;21(12):568–575.
  • Tsai HC, Lehman CW, Chen CM. Use of platelet-rich plasma and platelet-derived patches to treat chronic wounds. J Wound Care. 2019 Jan 2;28(1):15–21.
  • Mazzocca AD, McCarthy MB, Chowaniec DM, et al. Platelet-rich plasma differs according to preparation method and human variability. J Bone Joint Surg Am. 2012 Feb 15;94(4):308–316.
  • Patel S, Dhillon MS, Aggarwal S, et al. Treatment with platelet-rich plasma is more effective than placebo for knee osteoarthritis: a prospective, double-blind, randomized trial. Am J Sports Med. 2013;41(2):356–364.
  • Sam G, Vadakkekuttical RJ, Amol NV. In vitro evaluation of mechanical properties of platelet-rich fibrin membrane and scanning electron microscopic examination of its surface characteristics. J Indian Soc Periodontol. 2015;19(1):32–36. Jan-Feb.
  • Qiu M, Chen D, Shen C, et al. Platelet-Rich Plasma-Loaded Poly(d,l-lactide)-poly(ethylene glycol)-poly(d,l-lactide) hydrogel dressing promotes full-thickness skin wound healing in a rodent model. Int J Mol Sci. 2016 Jun 24;17(7):1001.
  • Mohammadi R, Mehrtash M, Mehrtash M, et al. Effect of platelet rich plasma combined with chitosan biodegradable film on full-thickness wound healing in rat model. Bull Emerg Trauma. 2016;4(1):29–37.
  • Kim W, Jang CH, Kim G. Optimally designed collagen/polycaprolactone biocomposites supplemented with controlled release of HA/TCP/rhBMP-2 and HA/TCP/PRP for hard tissue regeneration. Mater Sci Eng C Mater Biol Appl. 2017 Sep 1;78:763–772.
  • Liu J, Nie H, Xu Z, et al. Construction of PRP-containing nanofibrous scaffolds for controlled release and their application to cartilage regeneration. J Mater Chem B. 2015;3(4):581–591.
  • Lei X, Yang Y, Shan G, et al. Preparation of ADM/PRP freeze-dried dressing and effect of mice full-thickness skin defect model. Biomed Mater. 2019 Mar 7;14(3):035004.
  • Liu X, Yang Y, Niu X, et al. An in situ photocrosslinkable platelet rich plasma - complexed hydrogel glue with growth factor controlled release ability to promote cartilage defect repair. Acta Biomater. 2017 Oct 15;62:179–187.
  • Notodihardjo PV, Morimoto N, Kakudo N, et al. Gelatin hydrogel impregnated with platelet-rich plasma releasate promotes angiogenesis and wound healing in murine model. J Artif Organs. 2014 Oct 18;18(1);64-71.
  • Toffler M, Toscano N, Holtzclaw D, et al. Introducing Choukroun’s platelet rich fibrin (PRF) to the reconstructive surgery milieu. J Implant Adv Clin Dent. 2009;1(6):21–30.
  • Suzuki S, Morimoto N, Ikada Y. Gelatin gel as a carrier of platelet-derived growth factors. J Biomater Appl. 2013;28(4):595–606.
  • Sun H, Lv H, Qiu F, et al. Clinical application of a 3D-printed scaffold in chronic wound treatment: a case series. J Wound Care. 2018 May 2;27(5):262–271.
  • Barsotti MC, Losi P, Briganti E, et al. Effect of platelet lysate on human cells involved in different phases of wound healing. PLoS One. 2013;8(12):e84753.
  • Ranzato E, Patrone M, Mazzucco L, et al. Platelet lysate stimulates wound repair of HaCaT keratinocytes. Br J Dermatol. 2008;159(3):537–545.
  • Rossi S, Faccendini A, Bonferoni MC, et al. “Sponge-like” dressings based on biopolymers for the delivery of platelet lysate to skin chronic wounds. Int J Pharm. 2013 Jan 20;440(2):207–215.
  • Mori M, Rossi S, Ferrari F, et al. Sponge-like dressings based on the association of chitosan and sericin for the treatment of chronic skin ulcers. ii. loading of the hemoderivative platelet lysate. J Pharm Sci. 2016;105(3):1188–1195.
  • Nardini M, Perteghella S, Mastracci L, et al. Growth factors delivery system for skin regeneration: an advanced wound dressing. Pharmaceutics. 2020 Feb 3;12:2.
  • Sandri G, Bonferoni MC, Rossi S, et al. Platelet lysate formulations based on mucoadhesive polymers for the treatment of corneal lesions. J Pharm Pharmacol. 2011;63(2):189–198.
  • Sandri G, Bonferoni MC, Rossi S, et al. Platelet lysate and chondroitin sulfate loaded contact lenses to heal corneal lesions. Int J Pharm. 2016 Jul 25;509(1–2):188–196.
  • Sandri G, Bonferoni MC, Rossi S, et al. Thermosensitive eyedrops containing platelet lysate for the treatment of corneal ulcers. Int J Pharm. 2012 Apr 15;426(1–2):1–6.
  • Mori M, Rossi S, Bonferoni MC, et al. Calcium alginate particles for the combined delivery of platelet lysate and vancomycin hydrochloride in chronic skin ulcers. Int J Pharm. 2014 Jan 30;461(1–2):505–513.
  • Rossi S, Mori M, Vigani B, et al. A novel dressing for the combined delivery of platelet lysate and vancomycin hydrochloride to chronic skin ulcers: hyaluronic acid particles in alginate matrices. Eur J Pharm Sci. 2018 Jun 15;118:87–95.
  • Bonferoni MC, Sandri G, Rossi S, et al. Association of alpha tocopherol and Ag sulfadiazine chitosan oleate nanocarriers in bioactive dressings supporting platelet lysate application to skin wounds. Mar Drugs. 2018 Feb 9;16:2.
  • Laurens N, Koolwijk P, de Maat MP. Fibrin structure and wound healing. J Thromb Haemost. 2006;4(5):932–939.
  • Weisel JW, Litvinov RI. Fibrin formation, structure and properties. Subcell Biochem. 2017;82:405–456.
  • Heher P, Muhleder S, Mittermayr R, et al. Fibrin-based delivery strategies for acute and chronic wound healing. Adv Drug Deliv Rev. 2018;129:134–147.
  • Rajangam T, An SS. Fibrinogen and fibrin based micro and nano scaffolds incorporated with drugs, proteins, cells and genes for therapeutic biomedical applications. Int J Nanomed. 2013;8:3641–3662.
  • Whelan D, Caplice NM, Clover AJ. Fibrin as a delivery system in wound healing tissue engineering applications. J Control Release. 2014 Dec 28;196:1–8.
  • Drinnan CT, Zhang G, Alexander MA, et al. Multimodal release of transforming growth factor-beta1 and the BB isoform of platelet derived growth factor from PEGylated fibrin gels. J Control Release. 2010 Oct 15;147(2):180–186.
  • Ehrbar M, Djonov VG, Schnell C, et al. Cell-demanded liberation of VEGF121 from fibrin implants induces local and controlled blood vessel growth. Circ Res. 2004 Apr 30;94(8):1124–1132.
  • Martino MM, Hubbell JA. The 12th-14th type III repeats of fibronectin function as a highly promiscuous growth factor-binding domain. Faseb J. 2010;24(12):4711–4721.
  • Pandit AS, Feldman DS, Caulfield J, et al. Stimulation of angiogenesis by FGF-1 delivered through a modified fibrin scaffold. Growth Factors. 1998;15(2):113–123.
  • Geer DJ, Swartz DD, Andreadis ST. Biomimetic delivery of keratinocyte growth factor upon cellular demand for accelerated wound healing in vitro and in vivo. Am J Pathol. 2005;167(6):1575–1586.
  • Muhamed I, Sproul EP, Ligler FS, et al. Fibrin nanoparticles coupled with keratinocyte growth factor enhance the dermal wound-healing rate. ACS Appl Mater Interfaces. 2019 Jan 30;11(4):3771–3780.
  • Zhou W, Zhao M, Zhao Y, et al. A fibrin gel loaded with chitosan nanoparticles for local delivery of rhEGF: preparation and in vitro release studies. J Mater Sci Mater Med. 2011;22(5):1221–1230.
  • Wong C, Inman E, Spaethe R, et al. Fibrin-based biomaterials to deliver human growth factors. Thromb Haemost. 2003;89(3):573–582.
  • Briganti E, Spiller D, Mirtelli C, et al. A composite fibrin-based scaffold for controlled delivery of bioactive pro-angiogenetic growth factors. J Control Release. 2010 Feb 25;142(1):14–21.
  • Layman H, Li X, Nagar E, et al. Enhanced angiogenic efficacy through controlled and sustained delivery of FGF-2 and G-CSF from fibrin hydrogels containing ionic-albumin microspheres. J Biomater Sci Polym Ed. 2012;23(1–4):185–206.
  • Layman H, Rahnemai-Azar AA, Pham SM, et al. Synergistic angiogenic effect of codelivering fibroblast growth factor 2 and granulocyte-colony stimulating factor from fibrin scaffolds and bone marrow transplantation in critical limb ischemia. Tissue Eng Part A. 2011;17(1–2):243–254.
  • Mogford JE, Tawil B, Jia S, et al. Fibrin sealant combined with fibroblasts and platelet-derived growth factor enhance wound healing in excisional wounds. Wound Repair Regen. 2009;17(3):405–410.
  • Gwak SJ, Kim SS, Sung K, et al. Synergistic effect of keratinocyte transplantation and epidermal growth factor delivery on epidermal regeneration. Cell Transplant. 2005;14(10):809–817.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.